WO2013149026A2 - Utilisation de n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine en association avec des inhibiteurs d'histone désacétylase pour le traitement du cancer - Google Patents

Utilisation de n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine en association avec des inhibiteurs d'histone désacétylase pour le traitement du cancer Download PDF

Info

Publication number
WO2013149026A2
WO2013149026A2 PCT/US2013/034389 US2013034389W WO2013149026A2 WO 2013149026 A2 WO2013149026 A2 WO 2013149026A2 US 2013034389 W US2013034389 W US 2013034389W WO 2013149026 A2 WO2013149026 A2 WO 2013149026A2
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
amg
inhibiting agent
tumor
treatment
Prior art date
Application number
PCT/US2013/034389
Other languages
English (en)
Inventor
Michael Carducci
Sushant KACHHAP
Channing PALLER
Original Assignee
The Johns Hopkins University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Johns Hopkins University filed Critical The Johns Hopkins University
Priority to US14/388,949 priority Critical patent/US20150072988A1/en
Publication of WO2013149026A2 publication Critical patent/WO2013149026A2/fr
Priority to US15/088,799 priority patent/US20160213669A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/4045Indole-alkylamines; Amides thereof, e.g. serotonin, melatonin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/15Depsipeptides; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to the use of N-(4-((3-(2-amino-4-pyrimidinyl) -2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l-phthalazinamine in combination with histone deacetylase inhibitors (HDAC inhibitors) for the treatment of cancer.
  • HDAC inhibitors histone deacetylase inhibitors
  • Cancer is one of the most widespread diseases affecting Mankind, and a leading cause of death worldwide. In the United States alone, cancer is the second leading cause of death, surpassed only by heart disease. Cancer is often characterized by deregulation of normal cellular processes or unregulated cell proliferation. Cells that have been transformed to cancerous cells tend to proliferate in an uncontrolled and unregulated manner leading to, in some cases, metastisis or the spread of the cancer. Deregulation of the cell proliferation could result from the modification to one or more genes, responsible for the cellular pathways that control cell-cycle progression.
  • DNA modifications including but not limited to mutations, amplifications, rearrangements, deletions, and epigenetic gene silencing
  • one or more cell-cycle checkpoint regulators which allow the cell to move from one phase of the cell cycle to another unchecked.
  • mCRPC metastatic castration-resistant prostate cancer
  • docetaxel which arrests cells in prometaphase by binding to microtubules early in mitosis. It prevents cells from progressing to anaphase by forming multipolar spindles and maintaining activation of the spindle assembly checkpoint (Weaver BA, Cleveland DW. Decoding the links between mitosis, cancer, and chemotherapy: The mitotic checkpoint, adaptation, and cell death. Cancer Cell 2005;8:7-12).
  • Docetaxel causes severe side effects, including irreversible neuropathy, as microtubules in non-dividing cells are targeted as well. Furthermore, most tumors exhibit inherent resistance to docetaxel or acquire resistance during treatment
  • Histone deacetylases assist in the cell's control of the coiling and uncoiling of DNA around histones, in order to carry out gene expression.
  • the histone deacetylases accomplish this by acetylating the lysine residues in core histones leading to a less compact and more transcriptionally active chromatin.
  • HDAC histone deacetylases
  • HDAC inhibitors block this action and can result in
  • Histone deacetylase (HDAC) inhibitors are a new class of cytostatic agents that inhibit the proliferation of tumor cells in culture and in vivo by inducing cell cycle arrest, differentiation and/or apoptosis.
  • Histone acetylation and deacetylation play important roles in the modulation of chromatin topology and the regulation of gene transcription.
  • Histone deacetylase inhibition induces the accumulation of hyperacetylated nucleosome core histones in most regions of chromatin but affects the expression of only a small subset of genes, leading to transcriptional activation of some genes, but repression of an equal or larger number of other genes.
  • Non-histone proteins such as transcription factors are also targets for acetylation with varying functional effects.
  • Acetylation enhances the activity of some transcription factors such as the tumor suppressor p53 and the erythroid differentiation factor GATA-1 but may repress transcriptional activity of others including T cell factor and the co-activator ACTR.
  • ERalpha the estrogen receptor alpha
  • ACTR co-activator ACTR
  • HDAC inhibitors are classified based on their homology of accessory domains to yeast histone deacetylases.
  • the 18 currently known histone deacetylases are classified into four groups (groups I - IV): Class I, which includes HDAC-1, -2, -3 and -8 are related to yeasat RPD3 gene; Class II, which includes HDAC-4, -5, -6, -7, -9 and -10 are lrealted to yeast Hdal gene; Class III, also known as sirtuins are reated to the Sir2 gene and include SIRT1-7; and Class IV, which contains only HDAC 11 and has features of both Class I and Class II. (HDC Inhibitors Database, www.hdacis.com/index/html)
  • HDAC inhibitors are as follows.
  • Classical HDIs act exclusively on Class I and Class II HDACs by binding to the zinc-containing catalytic domain of the HDACs.
  • These classical HDIs fall into several groupings, in order of decreasing potency:
  • hydroxamic acids such as trichostatin A
  • cyclic tetrapeptides such as trapoxin B
  • depsipeptides
  • electrophilic ketones 4. electrophilic ketones; and 5. the aliphatic acid compounds such as phenylbutyrate and valproic acid.
  • “Second-generation” HDIs include the hydroxamic acids Vorinostat (SAHA), Belinostat (PXD101), LAQ824 (Drummond DC, Noble CO, Kirpotrin DB, Guo Z et al., Annu. Rev. Pharmacol. Toxicol. 45:495-528, 2005), and
  • Panobinostat (LBH589); and the benzamides: Entinostat (MS-275), CI994, and mocetinostat (MGCD0103) (Beckers T, Burkhardt C et al., Int. J. Cancer 121 (5): 1138- 1148, 2005 and Acharya MR, Sparreboom A. et al., Mol. Pharmacol 68 (4): 917-932, 2008).
  • the sirtuin Class III HDACs are dependent on NAD + and are, therefore, inhibited by nicotinamide, as well as derivatives of NAD, dihydrocoumarin, naphthopyranone, and 2-hydroxynaphaldehydes.
  • HDAC inhibitors have been shown to induce p21 (WAFl) expression, a regulator of p53's tumor suppressor activity.
  • WAFl p21
  • HDACs are involved in the pathway by which the retinoblastoma protein (pRb) suppresses cell proliferation.
  • the pRb protein is part of a complex that attracts HDACs to the chromatin so that it will deacetylate histones. Brehm A. et al., Nature 391 (6667): 597-601, 1998. HDAC1 negatively regulates the cardiovascular transcription factor Kruppel-like factor 5 through direct interaction. Matsumura T. et al., J. Biol. Chem. 280 (13): 12123-12129, 2005. Estrogen is well-established as a mitogenic factor implicated in the tumorigenesis and progression of breast cancer via its binding to the estrogen receptor alpha (ERa). Recent data indicate that chromatin inactivation mediated by HDAC and DNA methylation is a critical component of ERa silencing in human breast cancer cells. Zhang Z. et al., Breast Cancer Res. Treat. 94 (1): 11-16, 2005.
  • Figures 1-a and 1-b depicts the effects on cellular proliferation of prostate cancer cells in- vitro after treatment with AMG 900 and HDACs valproic acid or vorinostat, alone or in combination;
  • Figure 2 depicts the effects on expression of phospho-histone H3 in prostate cells in-vitro after treatment with AMG 900 and HDACs valproic acid or vorinostat, alone or in combination;
  • Figure 3 is a bar graph depicting the clonogenic survival in prostate cells in-vitro after combination treatment with AMG 900 and HDACs valproic acid or vorinostat;
  • Figure 4 is a bar graph depicting the effect on senescence in prostate cells in-vitro after combination treatment with AMG 900 and HDACs valproic acid or vorinostat;
  • Figure 5 depicts the effects on expression of p21 in prostate cells in-vitro after treatment with AMG 900 and HDACs valproic acid or vorinostat, alone or in combination;
  • Figure 6 is a graph depicting the in-vivo effects on tumor growth after treatment with AMG 900 and HDACs valproic acid or vorinostat, alone or in combination;
  • Figure 7 is a bar graph depicting the in-vivo effects on expression of phospho-histone H3 after treatment with AMG 900 and HDACs valproic acid or vorinostat, alone or in combination;
  • Figures 8A - 8D are bar graphs depicting the clonogenic survival in prostate cells in- vitro after combination treatment with AMG 900 and HDACs valproic acid or vorinostat.
  • the present invention provides for use of the compound, N-(4-((3-(2-amino-4- pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)- 1 -phthalazinamine (also referred to herein as "AMG 900" or “the compound”) or a pharmaceutically acceptable salt form thereof, in combination with an HDAC inhibitor for the treatment of cancer.
  • AMG 900 when administered with an HDAC inhibitor exhibited surprisingly superior and synergistic effects above what one of ordinary skill in the art would expect from treatment with either AMG 900 or the HDAC inhibitor alone.
  • AMG 900 has a chemical structure of:
  • the invention further provides use of a pharmaceutical composition comprising this compound, or a pharmaceutically acceptable salt form thereof, for therapeutic, prophylactic, acute or chronic treatment of cancer, including solid tumors of prostate cancer, breast cancer, ovarian cancer, lung cancer and the like, and other proliferating cancerous cells in patients.
  • the invention provides the use of AMG 900 in the manufacture of medicaments, and of pharmaceutical compositions, for the treatment of cancer in subjects who are in need of treatment, or who may have been previously treated solely with an HDAC inhibitor.
  • the invention further contemplates using AMG 900 in combination with such HDAC inhibitors presently approved for medical use by regulatory agencies, including without limitation Vorinostat and Romidepsin, as well as other HDAC agents undergoing clinical trials, including without limitation Panobinostat (LBH589), Valproic acid (as Mg valproate); Belinostat
  • the invention also includes use of any other pre -clinical HDAC inhibitors determined useful for the treatment of cancer via the reduction or inhibition of histone deacetylase activity in combination with AMG 900 for the treatment of cancer.
  • the invention provides a method of treating a solid tumor, including non-small cell lung cancer, breast cancer, and prostate cancer in a subject, the method comprising administering to the subject an effective dosage amount of AMG 900 or a
  • AMG 900 a small molecule pan Aurora A, B and C kinase inhibitor, has been found to provide a surprising and unexpected advantage in effect when use or administered in combination with a histone deacetylase (HDAC) inhibitor agent.
  • HDAC histone deacetylase
  • AMG 900 when administered, in dosages lower than when administered alone, in combination with an HDAC inhibitor exhibited positive and synergistic antiproliferative effects and long term clonogenic survival effects on cancer cells. Further, such combination treatments of tumors in-vivo exhibited effects on the growth curve of the tumor at least equivalent to, and superior than, that which was exhibited with a higher dosage of AMG 900 alone. Finally, the effects of the combination treatment were moderately synergistic to strongly synergistic compared to the single agent use, depending upon the concentrations used.
  • cancer refers to or describe the physiological condition in subjects that is typically characterized by unregulated cell growth.
  • examples of cancer include, without limitation, carcinoma, lymphoma, sarcoma, blastoma and leukemia. More particular examples of such cancers include squamous cell carcinoma, lung cancer, pancreatic cancer, cervical cancer, bladder cancer, hepatoma, breast cancer, colon carcinoma, and head and neck cancer. While the term “cancer” as used herein is not limited to any one specific form of the disease, it is believed that the combination treatment methods provided by the invention will be particularly effective for a variety of cancers in a subject in need of treatment.
  • HDAC inhibiting agent when used herein refers to a histone deacetylase inhibiting agent, which alone, may be useful for treating cancer. This term additionally refers to antineoplastic drugs used to treat cancer or a combination of these drugs into a standardized treatment regimen. Examples of HDAC inhibiting agents include, without limitation, member agents of Classes I - IV.
  • HDAC inhibiting agents include, without limitation, those which are approved for human use such as Vorinostat and Romidepsin, those which have been regulatorily approved for and are undergoing human clinical trials such as Panobinostat (LBH589; Ph III for various cancers constituing cutaneous T-cell lymphoma), valproic acid (administered as Mg valproate salt; in Ph III trials for cervical and ovarian cancer); Belinostat (PXD101; Ph II trial for relapsed ovarian cancer, and reportedly good results for T cell lymphoma), Mocetinostat (MGCD103; undergoing Ph II clinical trials for various cancers, including follicular lymphoma, Hodgkin lymphoma and acute myeloid leukemia (AML)),
  • Sulforaphane a novel histone deacetylase inhibitor in clinical trials as an anti-cancer agent.
  • the invention also includes use of any other pre-clinical HDAC inhibitors, including without limitation, Kevetrin, an agent selective for HDAC2, that are later determined to be useful for the treatment of cancer via the reduction or inhibition of histone deacetylase activity in combination with AMG 900.
  • the term “comprising” is meant to be open ended, including the indicated component(s) but not excluding other elements.
  • the term “refractory” when used here is intended to refer to not-yielding to, resistant or non-responsive to treatment, stimuli (therapy) or cure, including resistance to multiple therapeutic curative agents.
  • "Refractory” when used herein in the context of characterizing a cancer or tumor is intended to refer to the cancer or tumor being non- responsive or having a resistant or diminished response to treatment with one or more anticancer agents. The treatment typically is continual, prolonged and/or repetitive over a period of time resulting in the cancer or tumor developing resistance or becoming refractory to that very same treatment.
  • subject refers to any mammal, including humans and animals, such as cows, horses, dogs and cats.
  • the invention may be used in human patients as well as in veterinarian subjects and patients.
  • the subject is a human.
  • phrases "effective dosage amount” or “therapeutically-effective” when used in conjunction with AMG 900, or an HDAC inhibiting agent, is intended to quantify the amount of the compound (AMG 900 or HDAC inhibiting agent), which will achieve a reduction in size or severity of the cancer or tumor.
  • treat refers to therapy, including without limitation, curative therapy, prophylactic therapy, and preventative therapy.
  • Prophylactic treatment generally constitutes either preventing the onset of disorders altogether or delaying the onset of a pre-clinically evident stage of disorders in individuals.
  • pharmaceutically-acceptable salts embraces salts commonly used to form alkali metal salts and to form addition salts of free acids or free bases.
  • the nature of the salt is not critical, provided that it is pharmaceutically-acceptable.
  • Suitable pharmaceutically-acceptable acid addition salts of the compound may be prepared from an inorganic acid or from an organic acid. Examples of such inorganic acids include, without limitation, hydrochloric, hydrobromic, hydroiodic, nitric, carbonic, sulfuric and phosphoric acid.
  • organic acids include, without limitation, aliphatic, cycloaliphatic, aromatic, arylaliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which are formic, acetic, adipic, butyric, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, mesylic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, ethanedisulfonic, benzenesulfonic, pantothenic, 2-hydroxyethanesulfonic, toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, camphoric, camphorsulf
  • a bis-HCl, bis-sulfonic acid, bis-methanesulfonic acid, bis-benzenesulfonic acid, bis-toluenesulfonic acid, bis-aspartic acid, bis-malic acid or a bis-glutamic acid salt of AMG 900 is contemplated herein to be useful in combination with an HDAC inhibiting agent to treat cancer.
  • Suitable pharmaceutically-acceptable base addition salts of the compound include, without limitation, metallic salts such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or salts made from organic bases including primary, secondary, tertiary amines and substituted amines including cyclic amines such as caffeine, arginine, diethylamine, N-ethyl piperidine, aistidine, glucamine, isopropylamine, lysine, morpholine, N-ethyl morpholine, piperazine, piperidine, triethylamine, trimethylamine.
  • metallic salts such as salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc
  • organic bases including primary, secondary, tertiary amines and substituted amines including cyclic amines such as caffeine, arginine, diethylamine, N-ethyl piperidine, aistidine, glucamine, isopropylamine,
  • All of the salts contemplated herein may be prepared by conventional means from the corresponding compound by reacting, for example, the appropriate acid or base with the compound.
  • a base addition salt of a carboxylic acid or similar functional group on the HDAC inhibiting agent administered may be useful in carrying out the invention described herein.
  • Some useful HDAC inhibiting agents which may readily form base addition salts include, without limitation, hydroxamic acids or hydroxamates such as trichostatin A, vorinostat (also referred to herein as SAHA), belinostat and panobinostat, and aliphatic acid compounds such as phenyl butyrate and valproic acid.
  • AMG 900 N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4- methyl-2-thienyl)-l-phthalazinamine, may be prepared by the procedure analogous to that described in PCT publication WO2007087276, Example Methods Al or A2 on pg 70 but using l-chloro-4-(4-methyl-2-thienyl)phthalazine as the starting material, in conjunction with Examples 15 (pg 50), 25 (pg 55) and 30 (pg 59). These procedures are also described in US Patent No. 7,560,551, which specification is hereby incorporated herein by reference in its entirety. Specifically, AMG 900 may be prepared as described in Example 1 below.
  • Step 1 4-(2-chloropyridin-3-yl)pyrimidin-2-amine
  • Step 2 4-(2-(4-aminophenoxy)pyridin-3-yl)pyrimidin-2 -amine
  • Step 3 l -Chloro-4-(4-methylthiophen-2-yl)phthalazine 1 ,4-Dichlorophthalazine (1.40 g, 7.03 mmol), 4-methylthiophen-2-ylboronic acid (999 mg, 7.03 mmol), and PdCl 2 (DPPF) (721 mg, 985 ⁇ ) were added into a sealed tube. The tube was purged with Argon. Then sodium carbonate (2.0 M in water) (7.74 ml, 15.5 mmol) and 1 ,4-dioxane (35.2 ml, 7.03 mmol) were added.
  • DPPF PdCl 2
  • Step 4 N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2- thienvD-l -phthalazinamine
  • the mobile phase used a mixture of solvent A (H 2 O/0.1 % HO Ac) and solvent B (AcCN/0.1% HO Ac) with a 9 min time period for a gradient from 10% to 90% solvent B. The gradient was followed by a 0.5 min period to return to 10%o solvent B and a 2.5 min 10%> solvent B re -equilibration (flush) of the column.
  • solvent A H 2 O/0.1 % HO Ac
  • solvent B AcCN/0.1% HO Ac
  • Bodanszky A. Bodanszky, The Practice of Peptide Synthesis, Springer-Verlag, Berlin Heidelberg (1984); J. Seyden-Penne, Reductions by the Alumino- and Borohydrides in Organic Synthesis, 2 nd edition, Wiley-VCH, (1997); and L. Paquette, editor, Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995).
  • AMG 900 was tested for its ability to reduce or inhibit tumor progression in various cell lines (in-vitro) and multiple solid tumor types (in-vivo), some of which have previously been exposed to and developed resistance to standard-of-care antimitotic agents, including taxanes and vinca alkaloids, as well as to other chemotherapeutic agents.
  • the following Examples and resulting data will illustrate the synergistic ability of AMG 900, when used in combination with an HDAC inhibiting agent, to treat cancer. Unless otherwise indicated, the free base form of AMG 900 was used in the Examples described hereinbelow.
  • AMG 900-induced suppression of aurora kinase A and B activity inhibits cell proliferation in combination with an HDAC inhibiting agent such as VPA or SAHA
  • an HDAC inhibiting agent such as VPA or SAHA
  • the antiproliferative, expression of phosphor-histone H3 (pH3) and/or clonogenic survival effect of AMG 900 and an HDAC inihibiting agent, alone and in combination, were evaluated in vitro using prostate cancer cell lines.
  • Figures 1 -a, 1 -b, 2, 3, 4 and 5 AMG 900 exhibited useful and synergistic antiproliferative, cell survival and reduced downstream expression activity in prostate cancer cells. This anti cancer activity was seen with low concentrations and dosages of AMG 900.
  • AMG 900 in combination with an HDAC inhibiting agent, such as VPA or Vorinostat, provide the same or greater effect on cellular apoptosis and, therefore at treating cancer, than with a higher dose of AMG 900 as a single agent.
  • an HDAC inhibiting agent such as VPA or Vorinostat
  • proliferation activity of prostate cancer cells, DU-145, PC3 and LNCaP cell lines decreased after combination treatment with AMG 900 and HDACs VPA and vorinostat in a moderately synergistic manner at low concentrations in these cells.
  • AMG 900 was administered in combination with SAHA, it enhanced inhibition of cellular proliferation compared to single agent use in all three cell lines.
  • SAHA treatment combination exhibited synergistic effects in the PC3 cell lines, as illustrated in Figs 1-a and 1-b.
  • Figure 2 depicts the effects of the treatment of AMG 900 and HDACIs (HDAC Inhibitors) valproic acid or vorinostat, alone or in combination, on the expression of phospho -hi stone H3 in prostate cells.
  • Phosphorylated histone H3 (pH3), an indicator for aurora B activity and regarded as a biological marker for aurora kinase activity generally, is decreased both after AMG 900 and HDACIs treatment.
  • protein expression levels of pH3 are more decreased after combination treatment compared to single agent use.
  • Figure 3 is a bar graph which depicts the effects of the treatment of AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination, on the clonogenic survival in prostate cells.
  • Clonogenic assays indicate decreased clonogenic survival after combination treatment of low dose AMG 900 with VPA or SAHA. Depending on the concentrations used, the effect was moderately synergistic to strongly synergistic compared to single agent use.
  • Figure 4 is a bar graph which depicts the effect of the treatment of AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination, on senescence in prostate cells.
  • Ai represents AMG 900 alone at a 1 nM dose.
  • the HDACI VPA represents valproic acid alone while SAHA represents treatment with vorinostat alone.
  • the concentrations are also provided in the figure.
  • the precent positive cells in SA ⁇ -galactosidase assay indicates increased senescence after 48 hours of combination treatment of low dose AMG 900 with VPA or SAHA when compared to single agent use (p ⁇ 0.05), except for combination treatment of AMG 900 with SAHA in LNCaP cells. Imaging of the PC3 cells during the assay further confirmed this finding.
  • Figure 5 depicts the effect of the treatment of AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination, on expression of p21 in prostate cells.
  • AI represents AMG 900 at a low dose
  • A5 represents AMG 900 at a higher dose.
  • VI represents VPA alone while SI represents treatment with SAHA alone.
  • p21 a cyclin -dependent kinase inhibitor of cyclin-CDK2 and -4 complexes, increased in each cell line after combined treatment with AMG 900 and HDACIs. This may indicate increased arrest in Gl -phase, or simply increased stress in the cells at 48 hours.
  • FIGs 8A through 8D are bar graphs depicting the effects of the treatment of AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination, on the proliferation activity and long-term clonogenic survival in prostate cancer cells.
  • the graphs reveal additive and/or synergistic reduction in both proliferation activity and long term clonogenic survival after 48 hours of combination treatment of AMG 900 with VPA or SAHA.
  • Synergy was assessed using calcuSyn.
  • Ai is AMG 900; + represents moderate synergy; ++ represents good synergy; and +++ represents strong synergy. Depending on the concentrations used, the effect was moderately synergistic to strongly synergistic compared to single agent use.
  • Test article AMG 900
  • Prostate cancer cell lines (DU-145, LNCaP, PC3) were obtained from ATCC. Cells were grown in RPMI-1640 (Invitrogen) with 10% Fetal Bovine Serum (FBS)
  • MTS assays were performed with CellTiter 96TM Aqueous Non-Radioactive Cell Proliferation Assay reagent (Promega) according to the manufacturer's instructions.
  • prostate cancer cells were plated in 96-well plates, allowed to adhere overnight and treated with the compounds for 72 hours. Subsequently, the MTS reagent was added and after about two hours absorption at 490nm was measured using a colorimetric plate reader (Molecular Devices). Synergy was assessed using CalcuSyn software (BioSoft).
  • Clonogenic assays were performed to assess long term cell survival.
  • Prostate cancer cells were plated in complete RPMI media in 60 mm petri dishes. Upon reaching 50-60% confluency, drugs were added at the appropriate concentration and dishes were incubated for 48 hours. Then cells (1.25xl0 3 for DU-145 and PC3 cells, 2xl0 3 for LNCaP cells) were replated and grown in triplicate in new 60 mm dishes containing fresh, complete RPMI media. After 10-14 days, a crystal violet stain (Sigma) was administered to visualize colonies. Viable colonies were counted, and subsequently the presence and degree of synergistic effects were measured using CalcuSyn (Biosoft).
  • Prostate cancer cells were plated in 6-well plates (25-50x10 3 cells per well) and allowed to adhere overnight. Compounds were added at appropriate concentrations for 48 hours, after which senescent cells were stained using the senescence ⁇ -galactosidase staining kit (Cell Signaling Technology) according to the manufacturer's instructions. In brief, cells were washed in PBS and fixed in Fixative Solution (2% formaldehyde and 0.2% glutaraldehyde in IX PBS).
  • Fixative Solution 2% formaldehyde and 0.2% glutaraldehyde in IX PBS.
  • 5% milk in TBST (lOOmM Tris-HCl ph 7.4, 0.1% Tween20, 150mM NaCl in H 2 0) was used for p21 , cyclin B 1 , vinculin, cleaved PARP and all secondary antibodies; 5% BSA in TBST was used for the primary antibodies phosphorylated aurora, and phosphorylated and total H3. Blots were developed on film and scanned into a computer at 300 dots per inch (dpi). Densitometric analyses were performed using ImageJ (Research Services Branch, National Institute of Mental Health).
  • Dxl and Dx2 are the doses of drug 1 and drug 2 alone required to produce the same effect.
  • Synergism levels no synergism (CI>0.9 ), moderate synergism (0.7 ⁇ CI ⁇ 0.9, +), synergism (0.3 ⁇ CI ⁇ 0.7, ++), strong synergism (0.KCK0.3, +++), very strong synergism (CK0.1, ++++)) were determined from CI ranges, using the Chou-Talalay method according to the manufacturer's instructions. Chou TC. "Drug
  • AMG 900-induced suppression of aurora kinase A and B activity inhibits cell proliferation in combination with an HDAC inhibiting agent such as VPA or SAHA
  • an HDAC inhibiting agent such as VPA or SAHA
  • AMG 900 exhibited useful and synergistic effects on tumor growth and reduced downstream expression of phospho-histone H3 (pH3). This anti cancer activity was seen with low concentrations and dosages of AMG 900.
  • AMG 900 in combination with an HDAC inhibiting agent, such as VPA or Vorinostat, provide the same or greater effect on tumor growth and, therefore at treating cancer, than with a higher dose of AMG 900 as a single agent.
  • an HDAC inhibiting agent such as VPA or Vorinostat
  • Figure 6 is a graph depicting the in-vivo effects on tumor growth after treatment with AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination.
  • A3.75 alone represents AMG 900 at a dosage of 3.75 mg/kg weight of the subject, while A7.5 alone represents AMG 900 at a dosage of 7.5 mg/kg weight of the subject.
  • SAHA alone represents treatment with vorinostat alone. The combined treatments are so indicated by the plus (+) sign.
  • Figure 7 is a bar graph depicting the in-vivo effects on expression of phospho- histone H3 after treatment with AMG 900 and HDACIs valproic acid or vorinostat, alone or in combination.
  • mice used in this study were 8-week old JHU Oncology NOD/SCIDs and were housed under aseptic conditions on a 12 hour light-dark cycle with food and water provided ad lib. Each cage contained ⁇ 5 mice; cages were differentiated by treatment groups.
  • mice were divided into homogenous groups (8-9 per group) according to tumor size. Once tumors reached a volume of 100- 400 mm 3 , mice began receiving medications in accordance with their assigned treatment group. Medications were administered in four-day cycles per week for a total duration of four weeks. SAHA (50mg/kg) was administered once daily via intraperitoneal injections, on mornings of days 1 -4 of the dosing cycle. AMG 900 was administered through gavage in either low or high doses, depending on the treatment group, on days 1 and 2 of each dosing cycle.
  • mice were treated with vehicle (2% HPMC (hydroxylpropyl methyl cellulose) and 1% Tween80 in DI (deionized) water, pH 2.2 with MSA (methane sulfonic acid)), or AMG 900 at a concentration of 3.75mg/kg or 7.5mg/kg (provided weekly by AMGEN). Tumor measurements and mice body weights were obtained on the day preceding each dosing cycle (day 0/7), as well as the final day of dosing (day 4). Tumors were collected 26-35 days after start of the treatment, when tumor volume reached 100 mm 3 .
  • vehicle 2% HPMC (hydroxylpropyl methyl cellulose) and 1% Tween80 in DI (deionized) water, pH 2.2 with MSA (methane sulfonic acid)
  • AMG 900 at a concentration of 3.75mg/kg or 7.5mg/kg (provided weekly by AMGEN).
  • mice were cardiac perfused with 2% paraformaldehyde, after which tumors were excised, infiltrated in sucrose, embedded in OCT (Optimal Cutting Temperature) media and stored at -80°C.
  • Tissue sections were prepared for hematoxylin-eosin staining by fixing the tissue in formalin and embedding it in paraffin after excision of the tumor.
  • Sections (20 ⁇ ) were cut from frozen tissues mounted in OCT (Sakura Finetek) and mounted on microscopic slides (Fisher Scientific). Sections were blocked in blocking buffer (5% goat serum, 0.5% BSA, 0.1% Triton-X 100 and 0.01% azide in PBS) and probed overnight with the primary antibody phosphorylated histone H3 in a 1 : 100 dilution (cell signaling technology). Then the sections were washed in PBS containing 0.1 %> Triton-X 100 and probed overnight with Alexa-fluor conjugated secondary antibody anti-Rabbit 546 in a 1 :500 dilution (Molecular Probes, Invitrogen).
  • the in vivo data from the DU-145 xenograft model was analyzed with a random intercept hierarchical linear model.
  • the primary statistical outcome was tumor volume.
  • tumor volumes on days 3 through 26 were divided by the volume on day -1 and then the log of these values was taken for analysis.
  • the intercept in this longitudinal model was specified such that it represented the log ratio of the final tumor volume to the initial volume (time was coded using negative numbers and 0 for the final day).
  • y it denotes the log ratio tumor volume for mouse i at time t
  • ⁇ 0 is the intercept representing the log ratio of the day 26 tumor volume to the initial volume for the control group
  • ⁇ i is the linear effect of time for the control group
  • ⁇ 2 is the group effect on day 26,? 3 is the group effect in terms of the linear effect of time.
  • the mouse specific effect, ⁇ 0! represented the deviation of each mouse from the group intercept. This corrected for the correlation between measurements taken on the same mouse. Since the mice in this experiment were considered a representative sample from a larger population, the effect was considered random and it was assumed that the population distribution from which they were sampled had a normal distribution.
  • Tumor Volume (mm 3 ) Tumor Volume (mm 3 )
  • Tumor inhibition was calculated as follows: First; take [Initial tumor volume minus final tumor volume] for control and all treatment groups; second, take the change in treated tumor volume divided by control tumor volume, minus one and then multiply by 100.
  • Figure 6 hereinbelow describe the tumor volume and tumor growth results.
  • Combination treatment with AMG 900 and SAHA surprisingly resulted in synergistically significant tumor growth inhibition using both doses of AMG 900 compared with the single agent group and vehicle control group ( Figure 6).
  • in-vivo expression of the molecular marker pH3 was measured after AMG 900 and/or SAHA treatment, alone or in combination.
  • Figure 7 illustrates the results, which show that levels of pH3 are decreased in mice with DU-145 tumors after treatment with a low dose of AMG 900 in combination with treatment with SAHA.
  • Figure 7 further illustrates that the low dose effect is at least comparable, and maybe even superior, to single agent AMG 900 treatment at the higher dose.
  • AMG 900 is a pan aurora kinase inhibitor.
  • Aurora kinase proteins play a part in cell cycling and, therefore, cell proliferation.
  • Aurora kinases are enzymes of the serine/threonine kinase family of proteins, which play an important role in protein phosphorylation during the mitotic phase of the cell cycle.
  • Aurora A Aurora A
  • Aurora B Aurora C
  • Aurora 2 Aurora 1
  • Aurora 3 Aurora 3
  • Aurora-A is localized to the centrosome during interphase and is important for centrosome maturation and to maintain separation during spindle assembly.
  • Aurora-B localizes to the kinetochore in the G2 phase of the cell cycle until metaphase, and relocates to the midbody after anaphase.
  • Aurora-C was thought to function only in meiosis, but more recently has been found to be more closely related to Aurora-B, showing some overlapping functions and similar localization patterns in mitosis.
  • Each aurora kinase appears to share a common structure, including a highly conserved catalytic domain and a very short N-terminal domain that varies in size. (See R. Giet and C. Prigent, J. Cell. 112:3591-3601 (1999)).
  • Aurora kinases are over expressed in various types of cancers, including colon, breast, lung, pancrease, prostate, bladder, head, neck, cervix, and ovarian cancers.
  • the Aurora-A gene is part of an amplicon found in a subset of breast, colon, ovarian, liver, gastric and pancreatic tumors.
  • Aurora-B has also been found to be over expressed in most major tumor types. Over expression of Aurora-B in rodent fibroblasts induces transformation, suggesting that Aurora-B is oncogenic. More recently, Aurora-B mRNA expression has been linked to chromosomal instability in human breast cancer. (Y. Miyoshi et al., Int. J. Cancer, 92:370-373 (2001)).
  • HDAC inhibitors can induce p21 (WAF1) expression, a regulator of p53's tumor suppressor activity.
  • HDACs are involved in the pathway by which the retinoblastoma protein (pRb) suppresses cell proliferation.
  • the pRb protein is part of a complex that attracts HDACs to the chromatin so that it will deacetylate histones.
  • HDAC1 negatively regulates the cardiovascular transcription factor Kruppel- like factor 5 through direct interaction.
  • Estrogen is well-established as a mitogenic factor implicated in the tumorigenesis and progression of breast cancer via its
  • the present invention provides a method of treating cancer by administering an aurora kinase inhibitor compound, AMG 900, in combination with an HDAC inhibiting agent, such as presently approved HDACs for medical use by regulatory agencies, including without limitation, Vorinostat and Romidepsin, and HDAC agents in clinical trials, including without limitation Panobinostat (LBH589), Valproic acid (as Mg valproate); Belinostat (PXD101), Mocetinostat (MGCD103), Abexinostat (PCI-24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357), CUDC-101, AR-42, CHR-2845, CHR-3996, 4SC-202, CG200745, ACY-1215 and Sulforaphane.
  • the invention further provides AMG 900 in combination with other pre-clinical HDAC inhibitors in the treatment of cancer.
  • DU-145 and PC3 cells lines the low-dose AMG 900 (InM) combinations demonstrated equivalent clonogenic survival to high dose AMG 900 treatment (5nM), either alone or in combination with HDACIs. Morphologic changes were noted in treated cell lines and senescence was explored with beta-galactosidase staining.
  • a method of treating cancer in a subject comprising administering to the subject an effective dosage amount of AMG 900, or a pharmaceutically acceptable salt thereof, in combination with an HDAC inhibiting agent, wherein the combined therapy treats the cancer.
  • the HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXD101), Mocetinostat (MGCD103), Abexinostat (PCI- 24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357), CUDC-101, AR-42, CHR-2845, CHR-3996, 4SC-202, CG200745, ACY-1215 or Sulforphane.
  • the HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXD101), Mocetinostat (MGCD103), Abexinostat (PCI- 24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357), CUDC-101, AR-42, C
  • HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXD101), Mocetinostat (MGCD103), Abexinostat (PCI-24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357).
  • HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589) or valproic acid.
  • the cancer is one or more of (a) a solid or hematologically derived tumor selected from (a) cancer of the bladder, breast, colon, kidney, liver, lung, small cell lung cancer, esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate and skin, (b) a hematopoietic tumor of lymphoid lineage selected from leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell- lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma, (c) a hematopoietic tumor of myeloid lineage selected from acute and chronic myelogenous leukemias, myelodysplasia syndrome and promye
  • cancer is one or more of a solid tumor selected from cancer of the bladder, breast, colon, kidney, liver, lung, non-small cell lung, head and neck, esophageal, gastric, ovary, pancreas, stomach, cervix, thyroid and prostate or a lymphoma or leukemia.
  • any one of embodiments 1 -6 wherein the cancer is prostate cancer, ovarian cancer, breast cancer, cholangiocarcinoma, acute myeloid leukemia, chronic myeloid leukemia or a combination thereof.
  • the effective dosage amount of AMG 900 or a is provided.
  • pharmaceutically acceptable salt thereof is in the range of about 0.5 mg/kg to about 30 mg/kg weight of the subject.
  • pharmaceutically acceptable salt thereof is in the range of about 2.5 mg/kg to about 24 mg/kg weight of the subject.
  • pharmaceutically acceptable salt thereof is in the range of about 2.5 mg/kg to about 10 mg/kg weight of the subject.
  • a method of reducing the size of a solid tumor in a subject comprising administering to the subject an effective dosage amount of the compound AMG 900, or a pharmaceutically acceptable salt thereof, in combination with an HDAC inhibiting agent, wherein the combined therapy reduces the size of the tumor.
  • embodiment 15 of the invention there is provided a method of any one of embodiments 1-12 wherein the AMG 900, or a pharmaceutically acceptable salt thereof, and the HDAC inhibiting agent are administered sequentially or co-administered simultaneously.
  • embodiment 16 of the invention there is provided a method of any one of embodiments 1-12 wherein the AMG 900, or a pharmaceutically acceptable salt thereof, and the HDAC inhibiting agent are co-administered in a single dosage formulation.
  • embodiment 17 of the invention there is provided a method of any one of embodiments 1-12 wherein the AMG 900, or a pharmaceutically acceptable salt thereof, and the HDAC inhibiting agent are co-administered as separate dosage formulations.
  • HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXDIOI), Mocetinostat (MGCD103), Abexinostat (PCI-24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357), CUDC-101, AR-42, CHR-2845, CHR-3996, 4SC-202, CG200745,
  • HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXDIOI), Mocetinostat (MGCD103), Abexinostat (PCI-24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat (ITF2357), CUDC-101, AR-42, CHR-2845, CHR-3996, 4SC
  • the combination treatment of AMG 900 with an HDAC inhibiting agent is useful for, but not limited to, the prevention or treatment of cancer including, for example, various solid and hematologically derived tumors, such as carcinomas, including, without limitation, cancer of the bladder, breast, colon, kidney, liver, lung
  • hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias (AML and CML), myelodysplastic syndrome and promyelocytic leukemia); tumors of mesenchymal origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g.
  • tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma), where such cancers have relapsed or become refractory .
  • Cancers, such as prostate cancer, ovarian cancer, lung cancer, breast cancer, cholangiocarcinoma or other types of cancer, which have become refractory to anti-cancer treatment, such as with hormones, may also be treated with AMG 900.
  • AMG 900 can be used in combination with an HDAC inhibitor, such as vorinostat or valprioc acid, to treat breast cancer (Siegel D., J. Hematologic Oncology, 2:31, 2009).
  • HDAC inhibitor such as vorinostat or valprioc acid
  • Triple negative breast cancer is an aggressive form of breast cancer characterized by the lack of hormone receptors, estrogen and progesterone receptors, and HER2/neu expression, for which there are no effective treatment options.
  • the invention provides the method of any one of embodiments
  • cancer treated is a solid tumor selected from cancer of the bladder, breast, colon, kidney, liver, lung, non-small cell lung, head and neck, esophageal, gastric, ovary, pancreas, stomach, cervix, thyroid and prostate cancer.
  • the invention provides the method of any one of embodiments
  • the invention provides the method of any one of embodiments 1-17 wherein the HDAC inhibiting agent is Vorinostat, Romidepsin, Panobinostat (LBH589), valproic acid, Belinostat (PXD101), Mocetinostat (MGCD103), Abexinostat (PCI-24781), Entinostat (MS-275), SB939, Resminostat (4SC-210), Givinostat
  • the cancer treated is a solid tumor selected from cancer of the bladder, breast, colon, kidney, liver, lung, non-small cell lung, head and neck, esophageal, gastric, ovary, pancreas, stomach, cervix, thyroid and prostate cancer.
  • the invention provides the method of any one of embodiments 1-19 and 21 wherein the HDAC inhibiting agent is Vorinostat or valproic acid and the cancer treated is a solid tumor selected from cancer of the bladder, breast, colon, kidney, liver, lung, non-small cell lung, head and neck, esophageal, gastric, ovary, pancreas, stomach, cervix, thyroid and prostate cancer.
  • the HDAC inhibiting agent is Vorinostat or valproic acid
  • the cancer treated is a solid tumor selected from cancer of the bladder, breast, colon, kidney, liver, lung, non-small cell lung, head and neck, esophageal, gastric, ovary, pancreas, stomach, cervix, thyroid and prostate cancer.
  • the invention provides the method of any one of embodiments 1 -20 wherein the HDAC inhibiting agent is Vorinostat or valproic acid and the cancer treated is prostate cancer.
  • the invention provides the method of any one of embodiments 1-19 and 21-22 wherein the HDAC inhibiting agent is Vorinostat or valproic acid and the cancer treated is breast cancer.
  • the invention provides the method of any one of embodiments
  • HDAC inhibiting agent is Vorinostat or valproic acid and the cancer treated is triple negative breast cancer.
  • the invention also provides a method for the treatment of solid tumors, sarcomas (especially Ewing's sarcoma and osteosarcoma), retinoblastoma, rhabdomyosarcomas, neuroblastoma, hematopoietic malignancies, including leukemia and lymphoma, tumor- induced pleural or pericardial effusions, and malignant ascites.
  • sarcomas especially Ewing's sarcoma and osteosarcoma
  • retinoblastoma especially Ewing's sarcoma and osteosarcoma
  • rhabdomyosarcomas retinoblastoma
  • neuroblastoma hematopoietic malignancies
  • hematopoietic malignancies including leukemia and lymphoma
  • tumor- induced pleural or pericardial effusions tumor- induced pleural or pericardial effusions
  • the compound is also useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like.
  • animals including horses, dogs, and cats may be similarly treated with a combination of AMG 900 and HDAV inhibitors for cancers.
  • Each of AMG 900 and the HDAC inhibiting agent may be administered to the cancer subject in combination as a single or separate pharmaceutical compositions or medicaments, comprising the active pharmaceutical ingredient (API), ie., AMG 900 (N- (4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-l- phthalazinamine) and the HDAC inhibiting agent, in association with one or more nontoxic, pharmaceutically-acceptable carriers, diluents and/or adjuvants (collectively referred to herein as "excipient" materials).
  • AMG 900, or a pharmaceutically acceptable salt form thereof, and the HDAC inhibitor API can be processed in accordance with conventional methods of pharmacy to produce the medicinal and pharmaceutical compositions for administration to patients, including humans and other mammals.
  • the pharmaceutical composition may be administered to the subject by any suitable route, adapted to such a route, and in a dose effective for the refractory cancer treatment intended.
  • the composition, or API may, for example, be administered orally, mucosally, topically, rectally, pulmonarily such as by inhalation spray, or parentally including intravascularly, intravenously, intraperitoneally, subcutaneously,
  • the pharmaceutical composition may be in the form of, for example, a tablet, capsule, suspension or liquid.
  • the pharmaceutical composition is preferably made in the form of a dosage unit containing a particular amount of the active ingredient.
  • dosage units are tablets or capsules.
  • these may contain an amount of active ingredient from about 1 to 2000 mg, and typically from about 1 to 500 mg.
  • a suitable daily dose for a human or other mammal may vary widely depending on the condition of the patient and other factors, but, once again, can be determined using routine methods and practices.
  • the amount of the API (AMG 900) which is administered and the dosage regimen for treating the refractory cancer condition depends on a variety of factors, including the age, weight, sex and medical condition of the subject, the type of disease, the severity of the cancer, the route and frequency of administration, and the physical and chemical properties of AMG 900 or its particular form, including the specific salt form.
  • a dosage regimen may vary.
  • a daily dose of about 0.01 to 500 mg/kg, advantageously between about 0.01 and about 50 mg/kg, more advantageously about 0.1 and about 30 mg/kg and even more advantageously between about 0.1 mg/kg and about 25 mg/kg body weight may be appropriate.
  • the invention provides a method of treating cancer in a subject, the method comprising administering to the subject AMG 900 or a pharmaceutically acceptable salt thereof in an effective dosage amount in the range from about 0.5 mg/kg to about 25 mg/kg, wherein the subject's cancer is refractory to treatment with an anti-mitotic agent.
  • the invention provides a method of treating cancer in a subject, the method comprising administering to the subject AMG 900 or a pharmaceutically acceptable salt thereof in an effective dosage amount in the range from about 1.0 mg/kg to about 20 mg/kg, wherein the subject's cancer is refractory to treatment with standard of care chemotherapeutic agent, including an anti-mitotic agent.
  • the invention provides a method of treating cancer in a subject, the method comprising administering to the subject AMG 900 or a pharmaceutically acceptable salt thereof in an effective dosage amount in the range from about 3.0 mg/kg to about 15 mg/kg, wherein the subject's cancer is refractory to treatment with an anti-mitotic agent.
  • the daily dose can be administered in one to four doses per day.
  • AMG 900 may be combined with one or more adjuvants or "excipients" appropriate to the indicated route of administration. If administered on a per dose basis, AMG 900 may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, to form the final formulation. For example, AMG 900 and the excipient(s) may be tableted or encapsulated by known and accepted methods for convenient administration.
  • suitable formulations include, without limitation, pills, tablets, soft and hard-shell gel capsules, troches, orally-dissolvable forms and delayed or controlled-release formulations thereof.
  • capsule or tablet formulations may contain one or more controlled-release agents, such as
  • hydroxypropylmethyl cellulose as a dispersion with the API(s).
  • a topical preparation of the AMG 900 to the affected area two to four times a day.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, pastes, suspensions and the like) and drops suitable for administration to the eye, ear, or nose.
  • a suitable topical dose of the active ingredient is 0.1 mg to 150 mg administered one to four, preferably one or two times daily.
  • the API may comprise from 0.001 % to 10% w/w, e.g., from 1 % to 2% by weight of the formulation, although it may comprise as much as 10%> w/w, but preferably not more than 5% w/w, and more preferably from 0.1 %> to 1 %> of the formulation.
  • AMG 900 When formulated in an ointment, AMG 900 may be employed with either paraffmic or a water-miscible ointment base. Alternatively, it may be formulated in a cream with an oil-in-water cream base. If desired, the aqueous phase of the cream base may include, for example at least 30%> w/w of a polyhydric alcohol such as propylene glycol, butane-l ,3-diol, mannitol, sorbitol, glycerol, polyethylene glycol and mixtures thereof.
  • the topical formulation may desirably include a compound, which enhances absorption or penetration of the active ingredient through the skin or other affected areas. Examples of such dermal penetration enhancers include DMSO and related analogs.
  • AMG 900 can also be administered by transdermal device.
  • transdermal administration will be accomplished using a patch either of the reservoir and porous membrane type or of a solid matrix variety.
  • AMG 900 is delivered continuously from the reservoir or microcapsules through a membrane into the active agent permeable adhesive, which is in contact with the skin or mucosa of the recipient. If AMG 900 is absorbed through the skin, a controlled and predetermined flow of AMG 900 is administered to the recipient.
  • the encapsulating agent may also function as the membrane.
  • the oily phase of the emulsions may be constituted from known ingredients in a known manner. While the phase may comprise merely an emulsifier, it may comprise a mixture of at least one emulsifier with a fat, or an oil, or with both a fat and an oil.
  • a hydrophilic emulsifier is included together with a lipophilic emulsifier which acts as a stabilizer. It is also preferred to include both an oil and a fat. Together, the emulsifier(s) with or without stabilizer(s) make-up the so-called emulsifying wax, and the wax together with the oil and fat make up the so-called emulsifying ointment base, which forms the oily dispersed phase of the cream formulations.
  • Emulsifiers and emulsion stabilizers suitable for use in the formulation include, for example, Tween 60, Span 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate, sodium lauryl sulfate, glyceryl distearate alone or with a wax, or other materials well known in the art.
  • the choice of suitable oils or fats for the formulation is based on achieving the desired cosmetic properties, since the solubility of the API in most oils likely to be used in pharmaceutical emulsion formulations is very low.
  • the cream should preferably be a non-greasy, non-staining and washable product with suitable consistency to avoid leakage from tubes or other containers.
  • Straight or branched chain, mono- or dibasic alkyl esters such as di-isoadipate, isocetyl stearate, propylene glycol diester of coconut fatty acids, isopropyl myristate, decyl oleate, isopropyl palmitate, butyl stearate, 2-ethylhexyl palmitate or a blend of branched chain esters may be used. These may be used alone or in combination depending on the properties required. Alternatively, high melting point lipids such as white soft paraffin and/or liquid paraffin or other mineral oils can be used.
  • Formulations suitable for topical administration to the eye also include eye drops wherein the active ingredients are dissolved or suspended in suitable carrier, especially an aqueous solvent for AMG 900.
  • AMG 900 is preferably present in such formulations in a concentration of 0.5 to 20%, advantageously 0.5 to 10% and particularly about 1.5% w/w.
  • Formulations for parenteral administration may be in the form of aqueous or nonaqueous isotonic sterile injection solutions or suspensions. These solutions and suspensions may be prepared from sterile powders or granules using one or more of the carriers or diluents mentioned for use in the formulations for oral administration or by using other suitable dispersing or wetting agents and suspending agents.
  • AMG 900 may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well and widely known in the pharmaceutical art.
  • AMG 900 may also be administered by injection as a composition with suitable carriers including saline, dextrose, or water, or with cyclodextrin (ie. Captisol), cosolvent solubilization (ie. propylene glycol) or micellar solubilization (ie. Tween 80).
  • suitable carriers including saline, dextrose, or water, or with cyclodextrin (ie. Captisol), cosolvent solubilization (ie. propylene glycol) or micellar solubilization (ie. Tween 80).
  • the sterile injectible preparation may also be a sterile injectible solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed, including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • the pharmaceutical composition may be administered in the form of an aerosol or with an inhaler including dry powder aerosol.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable non-irritating excipient such as cocoa butter and polyethylene glycols that are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • compositions may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. Tablets and pills can additionally be prepared with enteric coatings. Such compositions may also comprise adjuvants, such as wetting, sweetening, flavoring, and perfuming agents.
  • the present invention provides AMG 900 in combination with an HDAC inhibitor for the treatment of cancer.
  • each of the AMG 900 and the HDAC inhibitor independently can be formulated as separate compositions, such as a tablet, capsule or injectable solution, which is individually administered simultaneously or sequentially at different times.
  • both API's can be formulated as a single formulation, such as a unit tablet, capsule or injectable solution comprising both API's, and co-administered in a substantially simultaneous manner as a single composition.
  • the invention includes a single capsule having a fixed ratio of both active agents or multiple, separate capsules for each agent.
  • co-administration in defining the use of AMG 900 in the present invention is intended to embrace both modes of administration of the drug combination, ie., either as a single formulation

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pain & Pain Management (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/US2013/034389 2012-03-30 2013-03-28 Utilisation de n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine en association avec des inhibiteurs d'histone désacétylase pour le traitement du cancer WO2013149026A2 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US14/388,949 US20150072988A1 (en) 2012-03-30 2013-03-28 Use of n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine in combination with histone deacetylase inhibitors for treatment of cancer
US15/088,799 US20160213669A1 (en) 2012-03-30 2016-04-01 Use of n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine in combination with histone deacetylase inhibitors for treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261618090P 2012-03-30 2012-03-30
US61/618,090 2012-03-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US14388949 A-371-Of-International 2014-09-29
US15/088,799 Continuation US20160213669A1 (en) 2012-03-30 2016-04-01 Use of n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine in combination with histone deacetylase inhibitors for treatment of cancer

Publications (1)

Publication Number Publication Date
WO2013149026A2 true WO2013149026A2 (fr) 2013-10-03

Family

ID=49261394

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/034389 WO2013149026A2 (fr) 2012-03-30 2013-03-28 Utilisation de n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine en association avec des inhibiteurs d'histone désacétylase pour le traitement du cancer

Country Status (1)

Country Link
WO (1) WO2013149026A2 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015084649A1 (fr) 2013-12-03 2015-06-11 Amgen Inc. Formes cristallines de sels pharmaceutiquement acceptables de la n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine et leurs utilisations
KR101818865B1 (ko) * 2016-10-26 2018-01-15 연세대학교 산학협력단 항암제 병용투여용 약학 조성물
KR20180045656A (ko) * 2016-10-26 2018-05-04 연세대학교 산학협력단 내성암의 내성 극복 또는 치료용 약학 조성물

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015084649A1 (fr) 2013-12-03 2015-06-11 Amgen Inc. Formes cristallines de sels pharmaceutiquement acceptables de la n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine et leurs utilisations
US10053452B2 (en) 2013-12-03 2018-08-21 Amgen Inc. Crystalline forms of N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine salts and uses thereof
KR101818865B1 (ko) * 2016-10-26 2018-01-15 연세대학교 산학협력단 항암제 병용투여용 약학 조성물
KR20180045656A (ko) * 2016-10-26 2018-05-04 연세대학교 산학협력단 내성암의 내성 극복 또는 치료용 약학 조성물
KR101878650B1 (ko) * 2016-10-26 2018-07-16 연세대학교 산학협력단 내성암의 내성 극복 또는 치료용 약학 조성물

Similar Documents

Publication Publication Date Title
US20160213669A1 (en) Use of n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phenyl)-4-(4-methyl-2-thienyl)-1-phthalazinamine in combination with histone deacetylase inhibitors for treatment of cancer
JP6974392B2 (ja) 転移性前立腺癌の治療
Tang et al. Therapeutic applications of histone deacetylase inhibitors in sarcoma
Lee et al. Mechanisms of resistance to histone deacetylase inhibitors
ES2863996T3 (es) Terapia de combinación para el tratamiento del cáncer
Yu et al. Inhibition of NF-κB results in anti-glioma activity and reduces temozolomide-induced chemoresistance by down-regulating MGMT gene expression
US8110550B2 (en) HDAC inhibitors and hormone targeted drugs for the treatment of cancer
AU2013343425A1 (en) Pharmaceutical combination comprising a B-Raf inhibitor and a histone deacetylase inhibitor and their use in the treatment of proliferative diseases
EP2818170B1 (fr) N-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridine yl) oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phthalazinamine à utiliser dans le traitement d'un cancer résistant aux agents antimitotiques
JP7123806B2 (ja) 静止細胞標的化およびegfr阻害剤を用いた新生物の処置のための組み合わせ
Takai et al. Human endometrial and ovarian cancer cells: histone deacetylase inhibitors exhibit antiproliferative activity, potently induce cell cycle arrest, and stimulate apoptosis
US11160770B2 (en) Compounds, compositions and methods for treating oxidative DNA damage disorders
Wan et al. Artesunate protects against surgery-induced knee arthrofibrosis by activating Beclin-1-mediated autophagy via inhibition of mTOR signaling
JP2019524748A (ja) がんの治療におけるエリブリン及びヒストンデアセチラーゼ阻害剤の使用
WO2020132259A1 (fr) Compositions et méthodes de traitement de cancers par administration d'un médicament associé à la phénothiazine qui active la protéine phosphatase 2a (pp2a) avec une activité inhibitrice réduite ciblée sur le récepteur de dopamine d2 et toxicité associée
JP5792185B2 (ja) 線維増殖性障害および癌からなる群から選択されるβ−カテニン媒介障害を治療するための医薬組成物
WO2012175973A1 (fr) Traitement combiné comprenant un inhibiteur de hdac6 et un inhibiteur de akt
Minjie et al. Targeting pancreatic cancer cells by a novel hydroxamate-based histone deacetylase (HDAC) inhibitor ST-3595
WO2013149026A2 (fr) Utilisation de n-(4-((3-(2-amino-4-pyrimidinyl)-2-pyridinyl)oxy)phényl)-4-(4-méthyl-2-thiényl)-1-phtalazinamine en association avec des inhibiteurs d'histone désacétylase pour le traitement du cancer
JP2019508460A (ja) 増殖性疾病の組み合せ療法
Lee et al. A novel imidazopyridine analogue as a phosphatidylinositol 3-kinase inhibitor against human breast cancer
RU2754131C1 (ru) Комбинированная терапия ингибитором ezh2
EP3512602A2 (fr) Inhibiteurs du facteur de choc thermique et utilisations de ceux-ci
WO2018232252A1 (fr) Méthodes de traitement de gliomes à l'aide d'un inhibiteur de stat3
US11433069B2 (en) Methods of use and pharmaceutical combinations comprising histone deacetylase inhibitors and JAK1/2 inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13770058

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 14388949

Country of ref document: US

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13770058

Country of ref document: EP

Kind code of ref document: A2